Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 228
Filter
1.
Food Funct ; 14(3): 1573-1583, 2023 Feb 06.
Article in English | MEDLINE | ID: mdl-36655918

ABSTRACT

Dietary oil composition determines the pathological processes of alcoholic fatty liver disease (AFLD). Oil rich in saturated fatty acids protects, whereas oil rich in polyunsaturated fatty acids aggravates the alcohol-induced liver injury. However, limited studies have been conducted to address how monounsaturated fatty acids (MUFAs) enriched oil controls the pathological development of AFLD. Therefore, this study was designed to evaluate the effect of MUFA-enriched extra virgin olive oil (OO) on AFLD. Twenty C57BL/6J mice were randomly allocated into four groups and fed modified Lieber-DeCarli liquid diets containing isocaloric maltose dextrin a non-alcohol or alcohol with corn oil and OO for four weeks. Dietary OO significantly exacerbated alcohol-induced liver dysfunction, evidenced by histological examinations and disturbed biochemical parameters. Dietary OO with alcohol decreased hormone-sensitive lipase (HSL), phosphorylated 5'-AMP-activated protein kinase (p-AMPK), and carnitine palmitoyltransferase-Iα (CPT1α) expression, and increased sterol regulatory element-binding protein-1c (SREBP-1c), diacylglycerol acyltransferase-2 (DGAT2), and very low-density lipoprotein receptor (VLDLR) expression in the liver. It also promoted the expression of hepatic interleukin-6 (IL-6) and hepatic tumour necrosis factor-alpha (TNF-α) at the transcriptional level. Additionally, adipose tissue lipolysis partially had an etiologic effect on alcohol-induced hepatic steatosis under OO pretreatment. In conclusion, MUFA-enriched OO exacerbated liver dysfunction in vivo. OO should be cautiously considered as a unique dietary oil source for individuals with AFLD.


Subject(s)
Fatty Liver, Alcoholic , Mice , Animals , Olive Oil/pharmacology , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Fatty Acids, Monounsaturated/metabolism , Mice, Inbred C57BL , Liver/metabolism , Ethanol/metabolism , Fatty Acids/metabolism , Corn Oil/metabolism
2.
Life Sci ; 310: 121064, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36220368

ABSTRACT

AIMS: This work investigated the effects of creatine supplementation on different pathways related to the pathogenesis of non-alcoholic fatty liver disease and alcoholic liver disease. MAIN METHODS: To induce alcoholic liver disease, male Swiss mice were divided into three groups: control, ethanol and ethanol supplemented with creatine. To induce non-alcoholic fatty liver disease, mice were divided into three groups: control, high-fat diet and high-fat diet supplemented with creatine. Each group consisted of eight animals. In both cases, creatine monohydrate was added to the diets (1 %; weight/vol). KEY FINDINGS: Creatine supplementation prevented high-fat diet-induced non-alcoholic fatty liver disease progression, demonstrated by attenuated liver fat accumulation and liver damage. On the other hand, when combined with ethanol, creatine supplementation up-regulated key genes related to ethanol metabolism, oxidative stress, inflammation and lipid synthesis, and exacerbated ethanol-induced liver steatosis and damage, demonstrated by increased liver fat accumulation and histopathological score, as well as elevated oxidative damage markers and inflammatory mediators. SIGNIFICANCE: Our results clearly demonstrated creatine supplementation exerts different outcomes in relation to non-alcoholic fatty liver disease and alcoholic liver disease, namely it protects against high-fat diet-induced non-alcoholic fatty liver disease but exacerbates ethanol-induced alcoholic liver disease. The exacerbating effects of the creatine and ethanol combination appear to be related to oxidative stress and inflammation-mediated up-regulation of ethanol metabolism.


Subject(s)
Fatty Liver, Alcoholic , Liver Diseases, Alcoholic , Non-alcoholic Fatty Liver Disease , Male , Mice , Animals , Non-alcoholic Fatty Liver Disease/prevention & control , Non-alcoholic Fatty Liver Disease/complications , Creatine/pharmacology , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/prevention & control , Liver/metabolism , Diet, High-Fat/adverse effects , Dietary Supplements , Liver Diseases, Alcoholic/pathology , Ethanol/toxicity , Ethanol/metabolism , Oxidative Stress , Inflammation/pathology
3.
Molecules ; 27(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35163881

ABSTRACT

Ampelopsis grossedentata (AG) is an ancient medicinal plant that is mainly distributed and used in southwest China. It exerts therapeutic effects, such as antioxidant, anti-diabetic, and anti-inflammatory activities, reductions in blood pressure and cholesterol and hepatoprotective effects. Researchers in China recently reported the anti-obesity effects of AG extract in diet-induced obese mice and rats. To verify these findings, we herein investigated the effects of AG extract and its principal compound, ampelopsin, in high-fat diet (HFD)- and alcohol diet-fed mice, olive oil-loaded mice, and differentiated 3T3-L1 cells. The results obtained showed that AG extract and ampelopsin significantly suppressed increases in the weights of body, livers and abdominal fat and also up-regulated the expression of carnitine palmitoyltransferase 1A in HFD-fed mice. In olive oil-loaded mice, AG extract and ampelopsin significantly attenuated increases in serum triglyceride (TG) levels. In differentiated 3T3-L1 cells, AG extract and ampelopsin promoted TG decomposition, which appeared to be attributed to the expression of hormone-sensitive lipase. In alcohol diet-fed mice, AG extract and ampelopsin reduced serum levels of ethanol, glutamic oxaloacetic transaminase (GOT), and glutamic pyruvic transaminase (GPT) and liver TG. An examination of metabolic enzyme expression patterns revealed that AG extract and ampelopsin mainly enhanced the expression of aldehyde dehydrogenase and suppressed that of cytochrome P450, family 2, subfamily e1. In conclusion, AG extract and ampelopsin suppressed diet-induced intestinal fat accumulation and reduced the risk of fatty liver associated with HFD and alcohol consumption.


Subject(s)
Anti-Obesity Agents/pharmacology , Diet, High-Fat , Fatty Liver, Alcoholic/drug therapy , Flavonoids/pharmacology , Non-alcoholic Fatty Liver Disease/drug therapy , Plant Extracts/pharmacology , Tea/chemistry , 3T3-L1 Cells , Adiposity , Animals , Antioxidants/pharmacology , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Fatty Liver, Alcoholic/pathology , Lipid Metabolism , Lipogenesis , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Obese , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Phytotherapy , Rats , Rats, Sprague-Dawley
4.
Int J Mol Sci ; 23(2)2022 Jan 11.
Article in English | MEDLINE | ID: mdl-35054960

ABSTRACT

Alcoholic liver disease (ALD) is characterized by the injury, inflammation, and scarring in the liver owing to excessive alcohol consumption. Currently, ALD is a leading cause for liver transplantation. Therefore, extensive studies (in vitro, in experimental ALD models and in humans) are needed to elucidate pathological features and pathogenic mechanisms underlying ALD. Notably, oxidative changes in the liver have been recognized as a signature trait of ALD. Progression of ALD is linked to the generation of highly reactive free radicals by reactions involving ethanol and its metabolites. Furthermore, hepatic oxidative stress promotes tissue injury and, in turn, stimulates inflammatory responses in the liver, forming a pathological loop that promotes the progression of ALD. Accordingly, accumulating further knowledge on the relationship between oxidative stress and inflammation may help establish a viable therapeutic approach for treating ALD.


Subject(s)
Biomarkers , Disease Susceptibility , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Hepatitis/complications , Hepatitis/metabolism , Oxidative Stress , Signal Transduction , Animals , Disease Susceptibility/immunology , Ethanol/adverse effects , Ethanol/metabolism , Fatty Liver/complications , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver, Alcoholic/pathology , Gene Expression Regulation , Hepatitis/etiology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Immunity, Innate , Metabolic Networks and Pathways , MicroRNAs/genetics , Oxidation-Reduction
5.
Molecules ; 26(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34834064

ABSTRACT

Alcohol is metabolized in liver. Chronic alcohol abuse results in alcohol-induced fatty liver and liver injury. Red quinoa (Chenopodium formosanum) was a traditional staple food for Taiwanese aborigines. Red quinoa bran (RQB) included strong anti-oxidative and anti-inflammatory polyphenolic compounds, but it was usually regarded as the agricultural waste. Therefore, this study is to investigate the effect of water and ethanol extraction products of RQB on the prevention of liquid alcoholic diet-induced acute liver injury in mice. The mice were given whole grain powder of red quinoa (RQ-P), RQB ethanol extract (RQB-E), RQB water extract (RQB-W), and rutin orally for 6 weeks, respectively. The results indicated that RQB-E, RQB-W, and rutin decreased alcoholic diet-induced activities of aspartate aminotransferase and alanine aminotransferase, and the levels of serum triglyceride, total cholesterol, and hepatic triglyceride. Hematoxylin and eosin staining of liver tissues showed that RQB-E and RQB-W reduced lipid droplet accumulation and liver injury. However, ethanol extraction process can gain high rutin and antioxidative agents contents from red quinoa, that showed strong effects in preventing alcoholic fatty liver disease and liver injury via increasing superoxide dismutase/catalase antioxidative system and repressing the expressions of fatty acid synthesis enzyme acetyl-CoA carboxylase.


Subject(s)
Antioxidants/therapeutic use , Chenopodium quinoa , Fatty Liver, Alcoholic/prevention & control , Plant Extracts/therapeutic use , Rutin/therapeutic use , Animals , Antioxidants/chemistry , Chenopodium quinoa/chemistry , Ethanol/adverse effects , Fatty Acids/metabolism , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Lipogenesis/drug effects , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Plant Extracts/chemistry , Rutin/chemistry
6.
Nutrients ; 13(11)2021 Nov 20.
Article in English | MEDLINE | ID: mdl-34836410

ABSTRACT

The aim of this study was to evaluate the therapeutic effects of chlorogenic acid (CGA) in rats with advanced alcoholic steatohepatitis. The rats were fed on a high-fat diet and gavaged with ethanol (4 g/kg) for 8 weeks. The livers of ethanol-treated rats showed steatosis; necrosis and mononuclear infiltration; and significant upregulation of the mRNA expression of the prooxidant (Cyp2e1, iNos), lipogenic (Srebp1, Acc), proinflammatory (Tlr4, Nf-κb, TnfA, Il-1B, and Il-6), and profibrogenic (TgfB, Col1, VegfA) genes. Simultaneously, a downregulation of level of Sod and Nrf2 was observed, which was accompanied by increased serum transaminase, TnfA, and serum and liver triglycerides levels. CGA administration (40 and 80 mg/kg, 8 weeks) to ethanol-fed group reduced the liver expression levels of Cyp2e1 and iNos, whereas it markedly enhanced the expression of Sod, Nrf2, and Ho-1. CGA at both doses downregulated the expressions of lipogenic, proinflammatory, and profibrogenic genes, while the expression of Tlr4 was lowered only after the higher dose of CGA. The higher dose of CGA efficiently prevented the progression of alcohol-induced steatosis and reduced inflammation through regulation of the expression of genes encoding the proteins involved in the Tlr4/Nf-κB signaling pathway and fibrosis. The study revealed hepatoprotective and anti-inflammatory effects of CGA through the regulation of expression of genes encoding Cyp2e1/Nrf2 involved in oxidative stress modulation. These results demonstrate CGA as a therapeutic candidate for the prevention and treatment of alcoholic steatohepatitis.


Subject(s)
Chlorogenic Acid/pharmacology , Fatty Liver, Alcoholic/drug therapy , Homeostasis/drug effects , Oxidation-Reduction/drug effects , Protective Agents/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Cytochrome P-450 CYP2E1/metabolism , Diet, High-Fat , Ethanol , Fatty Liver, Alcoholic/etiology , Inflammation , Lipogenesis/drug effects , Liver/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Rats , Signal Transduction/drug effects
7.
Hepatology ; 73(3): 952-967, 2021 03.
Article in English | MEDLINE | ID: mdl-32418220

ABSTRACT

BACKGROUND AND AIMS: Chronic alcohol consumption is accompanied by intestinal inflammation. However, little is known about how alterations to the intestinal immune system and sphingolipids contribute to the pathogenesis of alcohol-associated liver disease (ALD). APPROACH AND RESULTS: We used wild-type mice, retinoid-related orphan receptor gamma t (RORγt)-deficient mice, sphingosine kinase-deficient mice, and local gut anti-inflammatory, 5-aminosalicyclic acid-treated mice in a chronic-binge ethanol feeding model. Targeted lipidomics assessed the sphingolipids in gut and liver samples. Gut immune cell populations, the amounts of sphingolipids, and the level of liver injury were examined. Alcohol intake induces a pro-inflammatory shift in immune cell populations in the gut, including an increase in Th17 cells. Using RORγt-deficient mice, we found that Th17 cells are required for alcohol-associated gut inflammation and the development of ALD. Treatment with 5-aminosalicyclic acid decreases alcohol-induced liver injury and reverses gut inflammation by the suppression of CD4+ /RORγt+ /interleukin-17A+ cells. Increased Th17 cells were due to up-regulation of sphingosine kinase 1 activity and RORγt activation. We found that S1P/S1PR1 signaling is required for the development of Th17 cell-mediated ALD. Importantly, in vivo intervention blocking of S1P/S1PR1 signaling markedly attenuated alcohol-induced liver inflammation, steatosis, and damage. CONCLUSIONS: Gut inflammation is a functional alteration of immune cells in ALD. Reducing gut Th17 cells leads to reduced liver damage. S1P signaling was crucial in the pathogenesis of ALD in a Th17 cell-dependent manner. Furthermore, our findings suggest that compounds that reduce gut inflammation locally may represent a unique targeted approach in the treatment of ALD.


Subject(s)
Ethanol/adverse effects , Fatty Liver, Alcoholic/prevention & control , Lysophospholipids/pharmacology , Sphingosine/analogs & derivatives , Th17 Cells/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Disease Models, Animal , Fatty Liver, Alcoholic/etiology , Female , Intestines/cytology , Intestines/drug effects , Intestines/immunology , Male , Mesalamine/pharmacology , Mesalamine/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Sphingosine/pharmacology , Th17 Cells/drug effects
8.
Food Funct ; 12(1): 362-372, 2021 Jan 07.
Article in English | MEDLINE | ID: mdl-33325949

ABSTRACT

Excessive alcohol drinking and a high-fat diet (HFD) promote steatohepatitis in the comorbidity of NAFLD and AFLD. Taxifolin (TAX) is a rich dihydroxyflavone compound found in onions, milk thistle and Douglas fir. We aimed to explore the intervention mechanism of TAX on chronic steatohepatitis induced by HFD feeding plus acute ethanol binge. We established an in vivo model by HFD feeding plus a single dose of ethanol binge, and established an in vitro model by oleic acid or palmitic acid on HepG2 cells to induce lipid accumulation. TAX regulated lipid synthesis by inhibiting the expression of SREBP1 and upregulating the PPARγ level. In addition, TAX inhibited the expression of P2X7R, IL-1ß, and caspase-1. Moreover, TAX reduced the expression of caspase-1 activation; thereby inhibiting the recruitment of macrophages and neutrophils. TAX also improved the inflammatory response caused by caspase-1 activation in steatotic hepatocytes. TAX exhibited an inhibitory effect on lipid accumulation and caspase-1-related pyroptosis. Collectively, TAX has therapeutic potential as an intervention of steatohepatitis induced by alcohol combined with HFD and for preventing non-alcoholic fatty liver degeneration targeting caspase-1-dependent pyroptosis.


Subject(s)
Diet, High-Fat/adverse effects , Ethanol/adverse effects , Fatty Liver, Alcoholic/prevention & control , Pyroptosis/drug effects , Quercetin/analogs & derivatives , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Binge Drinking/complications , Cells, Cultured , Central Nervous System Depressants/adverse effects , Disease Models, Animal , Fatty Liver, Alcoholic/etiology , Humans , Male , Mice , Mice, Inbred ICR , Quercetin/pharmacology
9.
Transplantation ; 104(12): 2528-2537, 2020 12.
Article in English | MEDLINE | ID: mdl-33215899

ABSTRACT

BACKGROUND: In an experimental murine liver clamping model, we aimed to investigate the efficacy of real-time confocal microscopy (RCM) in assessing viability of steatotic livers in comparison to standard assessment tools, including histopathological evaluation. METHODS: C57Bl/6 mice were subjected to a methionine-choline-deficient diet causing nonalcoholic fatty liver disease or to Lieber DeCarli diet causing ethanol-induced liver injury. Untreated animals served as controls. Liver biopsies were analyzed following challenge with 45 min of warm ischemia time and either 4 h of reperfusion or 24 h of cold storage. Organ quality assessment was performed at defined time points by RCM, histological staining, measurement of serum alanine aminotransferase activity, and expression analyses of proinflammatory cytokines. Additionally, survival analysis was performed. RESULTS: Cold as well as warm ischemia time resulted in a significant decrease in cell viability when compared with naive livers as well as nonischemic-challenged steatotic livers (P < 0.05) as assessed by RCM. Furthermore, RCM revealed the actual cellular damage at early time points, while established methods including H&E-staining and serum transaminase profile failed. CONCLUSIONS: In a translational attempt, we demonstrate that RCM is a suitable diagnostic tool to obtain information about functional damage of the liver apart from standard approaches.


Subject(s)
Fatty Liver, Alcoholic/pathology , Liver/pathology , Microscopy, Confocal , Non-alcoholic Fatty Liver Disease/pathology , Animals , Biopsy , Choline Deficiency/complications , Cold Ischemia/adverse effects , Disease Models, Animal , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/genetics , Fatty Liver, Alcoholic/metabolism , Liver/metabolism , Male , Methionine/deficiency , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Predictive Value of Tests , Reperfusion/adverse effects , Reperfusion Injury/etiology , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Time Factors , Tissue Survival , Warm Ischemia/adverse effects
10.
JCI Insight ; 5(14)2020 07 23.
Article in English | MEDLINE | ID: mdl-32544093

ABSTRACT

Alcohol-associated liver disease is a spectrum of liver disorders with histopathological changes ranging from simple steatosis to steatohepatitis, cirrhosis, and hepatocellular carcinoma. Recent data suggest that chronic-plus-binge ethanol intake induces steatohepatitis by promoting release by hepatocytes of proinflammatory mitochondrial DNA-enriched (mtDNA-enriched) extracellular vesicles (EVs). The aim of the present study was to investigate the role of the stress kinase apoptosis signal-regulating kinase 1 (ASK1) and p38 mitogen-activated protein kinase (p38) in chronic-plus-binge ethanol-induced steatohepatitis and mtDNA-enriched EV release. Microarray analysis revealed the greatest hepatic upregulation of metallothionein 1 and 2 (Mt1/2), which encode 2 of the most potent antioxidant proteins. Genetic deletion of the Mt1 and Mt2 genes aggravated ethanol-induced liver injury, as evidenced by elevation of serum ALT, neutrophil infiltration, oxidative stress, and ASK1/p38 activation in the liver. Inhibition or genetic deletion of Ask1 or p38 ameliorated ethanol-induced liver injury, inflammation, ROS levels, and expression of phagocytic oxidase and ER stress markers in the liver. In addition, inhibition of ASK1 or p38 also attenuated ethanol-induced mtDNA-enriched EV secretion from hepatocytes. Taken together, these findings indicate that induction of hepatic mtDNA-enriched EVs by ethanol is dependent on ASK1 and p38, thereby promoting alcoholic steatohepatitis.


Subject(s)
Extracellular Vesicles/genetics , Fatty Liver, Alcoholic/genetics , Inflammation/genetics , MAP Kinase Kinase Kinase 5/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Alcoholism/complications , Alcoholism/genetics , Alcoholism/pathology , Alcohols/toxicity , Animals , Binge Drinking/complications , Binge Drinking/genetics , Binge Drinking/pathology , DNA, Mitochondrial/genetics , Disease Models, Animal , Extracellular Vesicles/drug effects , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/pathology , Hepatocytes/drug effects , Humans , Inflammation/etiology , Inflammation/pathology , Liver/drug effects , Matrix Metalloproteinase 14/genetics , Metallothionein/genetics , Mice , Signal Transduction/drug effects
11.
Chem Biol Interact ; 327: 109176, 2020 Aug 25.
Article in English | MEDLINE | ID: mdl-32534989

ABSTRACT

Alcoholic liver disease (ALD) is a progressively aggravated liver disease with high incidence in alcoholics. Ethanol-induced fat accumulation and the subsequent lipopolysaccharide (LPS)-driven inflammation bring liver from reversible steatosis, to irreversible hepatitis, fibrosis, cirrhosis, and even hepatocellular carcinoma. Peroxisome proliferator-activated receptor α (PPARα) is a member of the nuclear receptor superfamily of ligand-activated transcription factors and plays pivotal roles in the regulation of fatty acid homeostasis as well as the inflammation control in the liver. It has been well documented that PPARα activity and/or expression are downregulated in liver of mice exposed to ethanol, which is thought to be one of the prime contributors to ethanol-induced steatosis, hepatitis and fibrosis. This article summarizes the current evidences from in vitro and animal models for the critical roles of PPARα in the onset and progression of ALD. Importantly, it should be noted that the expression of PPARα in human liver is reported to be similar to that in mice, and PPARα expression is downregulated in the liver of patients with nonalcoholic fatty liver disease (NAFLD), a disease sharing many similarities with ALD. Therefore, clinical trials investigating the expression of PPARα in the liver of ALD patients and the efficacy of strong PPARα agonists for the prevention and treatment of ALD are warranted.


Subject(s)
Fatty Liver, Alcoholic/etiology , PPAR alpha/metabolism , Adiponectin/metabolism , Animals , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Down-Regulation , Ethanol , Fatty Liver, Alcoholic/metabolism , Humans , Inflammation/chemically induced , Inflammation/etiology , Inflammation/metabolism , Liver/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , PPAR alpha/agonists , Sterol Regulatory Element Binding Protein 1/metabolism
12.
Toxicol Mech Methods ; 30(6): 407-416, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32237978

ABSTRACT

Mitogen-activated protein kinases (MAPKs) are versatile proteins that have been suggested to be involved in the regulation of lipid metabolism. This study was designed to investigate the responses of MAPK signaling to chronic ethanol exposure in vivo and in vitro, and try to explore its role in the pathogenesis of alcoholic fatty liver (AFL). Mice were fed with Lieber-Decarli liquid diet (5% ethanol, w/v) for 4 weeks to induce fatty liver, and the chronological changes of MAPK phosphorylation were measured using western blotting. We found that chronic ethanol feeding led to accumulation of triglyceride (TG), decreased phosphorylation of MAPKs, decreased protein level of peroxisomal proliferator activation receptor α (PPARα), and increased protein expression of cytochrome P4502E1 (CYP2E1) in mice liver. In vitro study showed that overexpression of CYP2E1 blunted the response of MAPKs to ethanol, and MAPK phosphatase 1 (MKP-1) knockdown by siRNA led to upregulation of PPARα protein level. Lastly, epidermal growth factor (EGF), a well-known MAPK activator, significantly suppressed chronic ethanol-induced hepatic fat accumulation and decline of PPARα expression in mice liver. Collectively, MAPK suppression, possibly due to the activation of hepatic CYP2E1, may be involved in chronic ethanol-induced hepatic steatosis.


Subject(s)
Fatty Liver, Alcoholic/enzymology , Liver/enzymology , Mitogen-Activated Protein Kinases/metabolism , Animals , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Disease Models, Animal , Down-Regulation , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Ethanol , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/genetics , Fatty Liver, Alcoholic/pathology , Hep G2 Cells , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Liver/pathology , Male , Mice, Inbred ICR , PPAR alpha/genetics , PPAR alpha/metabolism , Phosphorylation , Signal Transduction
13.
United European Gastroenterol J ; 8(3): 321-331, 2020 04.
Article in English | MEDLINE | ID: mdl-32213023

ABSTRACT

BACKGROUND: There is conflicting evidence regarding reliability criteria for the controlled attenuation parameter (CAP; a marker for hepatic steatosis [HS]). Thus, we assessed the diagnostic performance of CAP according to different reliability criteria based on real-world data from an academic centre. METHODS: Patients undergoing measurement of CAP and liver biopsy (±6 months) at the Medical University of Vienna were included. HS was assessed according to SAF score. RESULTS: In total 319 patients were included. The main aetiologies were non-alcoholic fatty liver disease (NAFLD, n = 177, 55.5%), viral hepatitis (n = 49, 15.4%), and alcoholic liver disease (ALD, n = 29, 9.1%). Histological steatosis and fibrosis stages were: S0: 93 (29.2%), S1: 100 (31.3%), S2: 67 (21.0%), and S3: 59 (18.5%); F0/F1: 150 (47.0%), F2: 47 (14.7%), and F3/F4: 122 (48.3%). In the overall cohort, the area under the receiver operating characteristic curve (AUC) of CAP was 0.843 (95% confidence interval [CI]: 0.798-0.887) for diagnosing HS ≥ S1), 0.789 (95%CI: 0.740-0.839) for ≥S2, and 0.767 (95%CI: 0.712-0.823) for S3. CAP corrections as suggested by Karlas et al. did not improve the diagnostic performance. Importantly, the AUC of CAP for HS ≥ S1 was numerically highest in patients with CAP-IQR/median<0.10 or <0.20 (obtained in 37.9% and 74.9%), in whom CAP also had better diagnostic performance, as compared with patients not meeting these criteria. Moreover, it was substantially higher in 288 (90.3%) patients with CAP-IQR/median<0.3: 0.856 (95%CI: 0.809-0.903) vs. patients not meeting this criterion (0.530 [95%CI: 0.309-0.751]). In contrast, the previously suggested reliability criterion of CAP-IQR<40 dB/m was not associated with an improved diagnostic performance for HS≥S1 (0.866 [95%CI: 0.812-0.920] vs. 0.799 [95%CI: 0.717-0.881]) and was only obtained in 199 (62.4%) patients. CONCLUSION: CAP-IQR/median<0.1, <0.2, and <0.3 identify reliable measurements for diagnosing any hepatic steatosis (≥S1). Importantly, CAP-IQR/median<0.3 has a considerably higher applicability in clinical practice, as compared with the previously suggested CAP-IQR<40 dB/m criterion.


Subject(s)
Elasticity Imaging Techniques/methods , Fatty Liver, Alcoholic/diagnosis , Hepatitis, Viral, Human/complications , Liver Cirrhosis/diagnosis , Non-alcoholic Fatty Liver Disease/diagnosis , Adult , Biopsy , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/pathology , Female , Hepatitis, Viral, Human/pathology , Humans , Liver/diagnostic imaging , Liver/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/pathology , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Predictive Value of Tests , ROC Curve , Reproducibility of Results , Retrospective Studies , Severity of Illness Index
14.
Curr Opin Gastroenterol ; 36(3): 164-168, 2020 05.
Article in English | MEDLINE | ID: mdl-32073504

ABSTRACT

PURPOSE OF REVIEW: This review will describe the natural history of alcohol-related liver disease (ArLD) in light of recent data that have synthesized existing knowledge on this topic and described new research cohorts to improve our understanding of progression and outcomes in ArLD. RECENT FINDINGS: ArLD occurs after a threshold of alcohol consumption, but this threshold is lowered by the presence of comorbid factors of which obesity is the most common. The most common stage of ArLD is alcohol-related steatosis: this is associated with a low rate of progression to cirrhosis (3%/year) and nonliver-related morbidity is more likely (4 versus 1%/year). In contrast, alcohol-related steatohepatitis or cirrhosis is more dangerous with higher rates of both nonliver and liver-related mortality. Hepatocellular carcinoma occurs at approximately 3%/year amongst people with ArLD cirrhosis. SUMMARY: These data allow an understanding of ArLD to accurately counsel patients and also to guide public health policies. Awareness of the shortcomings of the available data, highlighted in a recent systematic review, will inform the design of further research in particular to describe the multiple interacting factors that may cause ArLD to regress or progress.


Subject(s)
Alcohol Drinking , Liver Diseases, Alcoholic , Alcohol Drinking/adverse effects , Alcohol Drinking/epidemiology , Alcoholism/complications , Alcoholism/epidemiology , Disease Progression , Fatty Liver, Alcoholic/etiology , Humans , Liver Cirrhosis/etiology , Liver Diseases, Alcoholic/epidemiology , Liver Diseases, Alcoholic/etiology
15.
Nutrients ; 12(1)2020 Jan 19.
Article in English | MEDLINE | ID: mdl-31963802

ABSTRACT

Excess alcohol consumption is a top risk factor for death and disability. Fatty liver will likely develop and the risk of liver disease increases. We have previously demonstrated that an essential amino acid supplement (EAAS) improved protein synthesis and reduced intrahepatic lipid in the elderly. The purpose of this exploratory pilot study was to initiate the evaluation of EAAS on intrahepatic lipid (IHL), body composition, and blood lipids in individuals with mild to moderate alcohol use disorder (AUD). Following consent, determination of eligibility, and medical screening, 25 participants (18 males at 38 ± 15 years/age and 7 females at 34 ± 18 years/age) were enrolled and randomly assigned to one of two dosages: a low dose (LD: 8 g of EAAS twice/day (BID)) or high dose (HD: 13 g of EAAS BID). Five of the twenty-five enrolled participants dropped out of the intervention. Both groups consumed the supplement BID for 4 weeks. Pre- and post-EAAS administration, IHL was determined using magnetic resonance imaging/spectroscopy, body composition was analyzed using dual-energy X-ray absorptiometry, and blood parameters were measured by LabCorp. T-tests were used for statistical analysis and considered significant at p < 0.05. While there was no significant change in IHL in the LD group, there was a significant 23% reduction in IHL in the HD group (p = 0.02). Fat mass, lean tissue mass, bone mineral content, and blood lipids were not altered. Post-EAAS phosphatidylethanol was elevated and remained unchanged in LD at 407 ± 141 ng/mL and HD at 429 ± 196 ng/mL, indicating chronic and excess alcohol consumption. The HD of the proprietary EAAS formulation consumed BID seemed to lower IHL in individuals with mild to moderate AUD. We suggest that further studies in a larger cohort be conducted to more completely address this important area of investigation.


Subject(s)
Alcohol Drinking/adverse effects , Amino Acids, Essential/administration & dosage , Dietary Supplements , Fatty Liver, Alcoholic/drug therapy , Lipids/blood , Liver/drug effects , Adult , Alaska , Amino Acids, Essential/adverse effects , Biomarkers/blood , Body Composition/drug effects , Dietary Supplements/adverse effects , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Female , Humans , Liver/metabolism , Male , Middle Aged , Pilot Projects , Time Factors , Treatment Outcome , Young Adult
16.
J Agric Food Chem ; 68(6): 1750-1759, 2020 Feb 12.
Article in English | MEDLINE | ID: mdl-31971384

ABSTRACT

Alcohol is a globally well-established cause of fatty liver disease (FLD). Increased salt consumption is associated with an increased prevalence of adipocyte hypertrophy and liver injury. In this study, high dietary salt potentiated chronic alcohol-induced hepatic damage. We explored the physiological mechanism of alcoholic FLD in the gastrointestinal tract. Male C57BL/6J mice (8-week-old) were fed a high-salt diet (HSD; 4% NaCl) with or without chronic ethanol (CE) for 1 month. The fecal microbiota, serum biochemical indices, intestinal permeability, level of liver damage, and liver mitochondria were evaluated. The HSD, CE, and their combination (HSDE) significantly changed the gut microbiota's structure, and the HSDE mice contained more probiotic species (e.g., Bifidobacterium and Lactobacillus). The serum aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase levels were increased, and the lipid was accumulated in the liver tissues in the CE, HSD, and HSDE groups, which indicated liver damage, especially in the HSDE group. The increased intestinal permeability and mitochondrial dysfunction in the liver cells caused greater injury in the HSDE group than in the other groups. Thus, consuming HSD with alcohol contributes to FLD development and progression.


Subject(s)
Alcohol Drinking/adverse effects , Fatty Liver, Alcoholic/microbiology , Gastrointestinal Microbiome/drug effects , Sodium Chloride, Dietary/metabolism , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Ethanol/adverse effects , Ethanol/metabolism , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Feces/microbiology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Humans , Male , Mice , Mice, Inbred C57BL , Sodium Chloride, Dietary/adverse effects
17.
J Hepatol ; 72(4): 636-642, 2020 04.
Article in English | MEDLINE | ID: mdl-31954208

ABSTRACT

BACKGROUND & AIMS: Patients with alcoholic hepatitis and a modified Maddrey's discriminant function (mDF) <32 have a low risk of short-term mortality. However, few data exist concerning long-term outcomes. The aims of this study were to evaluate 5-year survival rates and to identify predictive factors for long-term prognosis in this patient population. METHODS: We studied patients from 2 centers who were admitted for hepatic decompensation (ascites, hepatic encephalopathy, or jaundice) and who had histological findings of steatohepatitis and an mDF <32. Clinical and biological parameters were recorded at the time of liver biopsy and alcohol consumption was recorded during follow-up. We performed Cox proportional hazard survival analysis to identify factors associated with 5-year survival. RESULTS: One hundred and twenty-one patients were included (male: 64%, mean age: 51.5 ± 10.3 years, presence of cirrhosis: 84%). The median model for end-stage liver disease and mDF scores were 14 (IQR 11.7-16.1) and 19 (IQR 11.1-24), respectively. During follow-up, 30% of the patients remained abstinent. Survival rates at 1, 6, 12, 24, and 60 months were 96.7 ± 1.6%, 90.1 ± 2.7%, 80.8 ± 3.6%, 69.9 ± 4.3%, and 50.7 ± 4.9%, respectively. The majority of deaths (80%) were liver related. In multivariable analysis, encephalopathy at baseline and alcohol abstinence were predictive of 5-year survival. The 5-year survival rates of patients without and with encephalopathy at baseline were 60.5 ± 5.8% and 29.7 ± 8.0%, respectively, and the 5-year survival rates of abstinent and non-abstinent patients were 74.0 ± 8.0% and 40.9 ± 5.8%, respectively. CONCLUSIONS: The mortality rate of patients with alcoholic hepatitis and an mDF <32 is around 50% at 5 years. Hepatic encephalopathy at baseline and lack of alcohol abstinence impair long-term prognosis. New treatment strategies, including measures to ensure abstinence, are required. LAY SUMMARY: Patients with alcoholic hepatitis that is of intermediate severity have a low risk of short-term mortality but not much is known regarding long-term outcomes for these patients. This study clearly indicates that patients with intermediate disease characteristics have poor long-term outcomes. The presence of hepatic encephalopathy at the time of diagnosis and the absence of alcohol abstinence during follow-up are factors that predict poor long-term mortality.


Subject(s)
End Stage Liver Disease/mortality , Fatty Liver, Alcoholic/mortality , Hepatic Encephalopathy/mortality , Hepatitis, Alcoholic/mortality , Liver Cirrhosis, Alcoholic/mortality , Severity of Illness Index , Adult , Aged , Alcohol Abstinence , Alcohol Drinking/adverse effects , End Stage Liver Disease/etiology , Fatty Liver, Alcoholic/etiology , Female , Follow-Up Studies , Hepatic Encephalopathy/etiology , Hepatitis, Alcoholic/etiology , Humans , Kaplan-Meier Estimate , Liver Cirrhosis, Alcoholic/etiology , Male , Middle Aged , Prognosis , Prospective Studies , Retrospective Studies , Survival Rate
18.
Am J Physiol Gastrointest Liver Physiol ; 318(3): G410-G418, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31905026

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD) are common causes of chronic liver disease. The overlap between ALD and NAFLD suggests the existence of metabolic steatohepatitis. Development of in vivo models that reflect various aspects of human steatohepatitis is essential for drug discovery. We aimed to characterize several models of steatohepatitis (SH) and to investigate whether the pathology could be modulated. Sprague-Dawley rats were fed a high-fat diet (HFD) for 9 wk, followed by either a high-fat, high-cholesterol and cholate diet (HFC) or a HFC diet containing 13% trans fat (HFC-TF). A subset received 15% ethanol-water twice a week for 12 wk. Serum triglycerides, cholesterol, LDL, HDL, aspartate aminotransferase (AST), alanine aminotransferase (ALT), and rodent NH2-terminal propeptide of type III collagen (rPRO-C3) were assessed. The liver was weighed and evaluated using modified Nonalcoholic Steatohepatitis Clinical Research Network histological score system criteria. All diets induced hepatomegaly, but only HFC-TF increased the size of visceral adipose tissue. Trans fat augmented HFC-induced dyslipidemia, and cholesterol was higher and HDL was lower in the HFC-TF groups. Alcohol lowered triglycerides in both dietary groups. HFC elevated ALT and AST, which were lowered by trans fat. All diets induced histological SH, addition of trans fat induced more steatosis but less inflammation. Inclusion of alcohol augmented the HFC-induced inflammation. All diets induced mild fibrosis. Inclusion of trans fat and alcohol significantly increased rPRO-C3. The addition of trans fat reduced the HFC-induced inflammation but augmented steatosis and dyslipidemia. Inclusion of alcohol induced a more inflammatory and fibrogenic phenotype.NEW & NOTEWORTHY Alcoholic liver disease and nonalcoholic liver disease share significant overlap, which suggests the existence of metabolic steatohepatitis. Trans fat has been implicated in steatohepatitis development. Here, we show that the addition of trans fat to an atherogenic diet results in a more steatotic but less inflammatory phenotype, whereas the addition of alcohol to an atherogenic diet augments the inflammatory and fibrogenic properties of the diet.


Subject(s)
Binge Drinking/complications , Diet, Atherogenic , Fatty Liver, Alcoholic/etiology , Liver/pathology , Non-alcoholic Fatty Liver Disease/etiology , Trans Fatty Acids , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Biomarkers/blood , Disease Models, Animal , Dyslipidemias/etiology , Dyslipidemias/metabolism , Dyslipidemias/pathology , Fatty Liver, Alcoholic/metabolism , Fatty Liver, Alcoholic/pathology , Hepatomegaly/etiology , Hepatomegaly/metabolism , Hepatomegaly/pathology , Lipid Metabolism , Liver/metabolism , Liver Cirrhosis, Alcoholic/etiology , Liver Cirrhosis, Alcoholic/metabolism , Liver Cirrhosis, Alcoholic/pathology , Male , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Oxidative Stress , Peptide Fragments/metabolism , Procollagen/metabolism , Rats, Sprague-Dawley
19.
Am J Physiol Gastrointest Liver Physiol ; 317(2): G182-G194, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31188634

ABSTRACT

Metabolic and alcoholic liver injuries result in nonalcoholic (NAFLD) or alcoholic (ALD) fatty liver disease, respectively. In particular, presence of fibrosis in NAFLD and ALD requires treatment, but development of drugs is hampered by the lack of suitable models with significant fibrosis. The carbon tetrachloride (CCl4) liver fibrosis model does not reflect human NAFLD or ALD, but CCl4 may serve as a fibrosis accelerator in addition to another injury. Ethanol in drinking water (16%) or Western diet (WD) were administered for 7 wk in mice either alone or in combination with CCl4 intoxications. Extent of fibrosis, steatosis, and inflammation was assessed by histology, transcription, and biochemistry. Furthermore, transcription of fibrosis, proliferation, and inflammation-related genes was studied on human liver samples with fibrosis resulting from hepatitis C virus infection (n = 7), NAFLD (n = 8), or ALD (n = 7). WD or ethanol alone induced only mild steatosis and inflammation. Combination of CCl4 and WD induced the most severe steatosis together with significant liver fibrosis and moderate inflammation. Combination of CCl4 and ethanol induced the strongest inflammation, with significant liver fibrosis and moderate steatosis. The relationship pattern between fibrosis, proliferation, and inflammation of human ALD was mostly similar in mice treated with CCl4 and ethanol. The combination of CCl4 intoxication with WD validates previous data suggesting it as an appropriate model for human nonalcoholic steatohepatitis. Especially, CCl4 plus ethanol for 7 wk induces ALD in mice, providing a model suitable for further basic research and drug testing.NEW & NOTEWORTHY Alcoholic fatty liver disease with significant fibrosis is generated within 7 wk using carbon tetrachloride as a fibrosis accelerator and administering gradually ethanol (up to 16%) in mice. The similarity in the pattern of steatosis, inflammation, and fibrosis involved in alcoholic fatty liver disease to those of the human condition renders this mouse model suitable as a preclinical model for drug development.


Subject(s)
Carbon Tetrachloride , Ethanol/metabolism , Fatty Liver, Alcoholic , Fatty Liver , Liver Cirrhosis , Animals , Carbon Tetrachloride/metabolism , Carbon Tetrachloride/toxicity , Disease Models, Animal , Fatty Liver/chemically induced , Fatty Liver/metabolism , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Humans , Inflammation/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Mice , Solvents/metabolism , Solvents/toxicity
20.
Am J Pathol ; 189(9): 1744-1762, 2019 09.
Article in English | MEDLINE | ID: mdl-31199920

ABSTRACT

Alcoholic fatty liver disease is often complicated by other pathologic insults, such as viral infection or high-fat diet. Autophagy plays a homeostatic role in the liver but can be compromised by alcohol, high-fat diet, or viral infection, which in turn affects the disease process caused by these etiologies. To understand the full impact of autophagy modulation on alcohol-induced liver injury, several genetic models of autophagy deficiency, which have different levels of functional alterations, were examined after acute binge or chronic-plus-binge treatment. Mice given alcohol with either mode and induced with deficiency in liver-specific Atg7 shortly after the induction of Atg7 deletion had elevated liver injury, indicating the protective role of autophagy. Constitutive hepatic Atg7-deficient mice, in which Atg7 was deleted in embryos, were more susceptible with chronic-plus-binge but not with acute alcohol treatment. Constitutive hepatic Atg5-deficient mice, in which Atg5 was deleted in embryos, were more susceptible with acute alcohol treatment, but liver injury was unexpectedly improved with the chronic-plus-binge regimen. A prolonged autophagy deficiency may complicate the hepatic response to alcohol treatment, likely in part due to endogenous liver injury. The complexity of the relationship between autophagy deficiency and alcohol-induced liver injury can thus be affected by the timing of autophagy dysfunction, the exact autophagy gene being affected, and the alcohol treatment regimen.


Subject(s)
Autophagy-Related Protein 7/physiology , Autophagy , Central Nervous System Depressants/toxicity , Chemical and Drug Induced Liver Injury/etiology , Ethanol/toxicity , Fatty Liver, Alcoholic/etiology , Animals , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Fatty Liver, Alcoholic/metabolism , Fatty Liver, Alcoholic/pathology , Female , Male , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...